51
|
Wu H, Wang Y, Yao Q, Fan L, Meng L, Zheng N, Li H, Wang J. Alkaline phosphatase attenuates LPS-induced liver injury by regulating the miR-146a-related inflammatory pathway. Int Immunopharmacol 2021; 101:108149. [PMID: 34634739 DOI: 10.1016/j.intimp.2021.108149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/09/2021] [Accepted: 09/07/2021] [Indexed: 11/27/2022]
Abstract
Lipopolysaccharide (LPS) can remain in dairy products after the sterilization of milk powder and may pose a threat to the health of infants and young children. There is a large amount of alkaline phosphatase (ALP) in raw milk, which can remove the phosphate bond of LPS, thus, detoxifying it. ALP is regarded as an indicator of the success of milk sterilization due to its strong heat resistance. ALP can alleviate the toxicity of LPS in enteritis and nephritis models, but the mechanism by which oral-intake of ALP protects liver tissue from LPS stimulation is unclear. In this study, an in vivo acute mouse liver injury model was induced by C. sakazakii LPS (200 μg/kg) and used to verify the protective mechanism of ALP (200 U/kg) on mice livers. The related pathways were also verified by in vitro cell culture. Enzyme linked immunosorbent assays (ELISAs), quantitative reverse transcription PCR (RT-qPCR) and western blotting were used to detect the levels of inflammatory factors at the protein level and RNA level, and to confirm the inflammation of liver tissue caused by LPS. ALP was found to alleviate acute liver injury in vitro by activating miR-146a. We found that ALP could up-regulate the level of miR146a and subsequently alleviates the expression of TLR4, TNF-α, matured IL-1β, and NF-κB in mouse liver tissue and hepatocytes; thus, reducing liver inflammation. Herein, we demonstrated for the first time that oral-intake of ALP protected liver tissue by up-regulating the expression of miR-146a and alleviating inflammatory reactions; thus, providing a research basis for the proper processing of milk. This study also suggests that producers should improve the awareness of the protective effects of bioactive proteins in raw milk.
Collapse
Affiliation(s)
- Haoming Wu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qianqian Yao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Linlin Fan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lu Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Huiying Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
52
|
Briolay A, Bessueille L, Magne D. TNAP: A New Multitask Enzyme in Energy Metabolism. Int J Mol Sci 2021; 22:ijms221910470. [PMID: 34638808 PMCID: PMC8509042 DOI: 10.3390/ijms221910470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is mainly known for its necessary role in skeletal and dental mineralization, which relies on the hydrolysis of the mineralization inhibitor inorganic pyrophosphate (PPi). Mutations in the gene encoding TNAP leading to severe hypophosphatasia result in strongly reduced mineralization and perinatal death. Fortunately, the relatively recent development of a recombinant TNAP with a bone anchor has allowed to correct the bone defects and prolong the life of affected babies and children. Researches on TNAP must however not be slowed down, because accumulating evidence indicates that TNAP activation in individuals with metabolic syndrome (MetS) is associated with enhanced cardiovascular mortality, presumably in relation with cardiovascular calcification. On the other hand, TNAP appears to be necessary to prevent the development of steatohepatitis in mice, suggesting that TNAP plays protective roles. The aim of the present review is to highlight the known or suspected functions of TNAP in energy metabolism that may be associated with the development of MetS. The location of TNAP in liver and its function in bile excretion, lipopolysaccharide (LPS) detoxification and fatty acid transport will be presented. The expression and function of TNAP in adipocyte differentiation and thermogenesis will also be discussed. Given that TNAP is a tissue- and substrate-nonspecific phosphatase, we believe that it exerts several crucial pathophysiological functions that are just beginning to be discovered.
Collapse
|
53
|
Huo JY, Jiang WY, Yin T, Xu H, Lyu YT, Chen YY, Chen M, Geng J, Jiang ZX, Shan QJ. Intestinal Barrier Dysfunction Exacerbates Neuroinflammation via the TLR4 Pathway in Mice With Heart Failure. Front Physiol 2021; 12:712338. [PMID: 34421655 PMCID: PMC8378453 DOI: 10.3389/fphys.2021.712338] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022] Open
Abstract
Aims The present study aimed to investigate alterations in neuroinflammation after heart failure (HF) and explore the potential mechanisms. Methods Male wild-type (WT) and Toll-like receptor 4 (TLR4)-knockout (KO) mice were subjected to sham operation or ligation of the left anterior descending coronary artery to induce HF. 8 weeks later, cardiac functions were analyzed by echocardiography, and intestinal barrier functions were examined by measuring tight junction protein expression, intestinal permeability and plasma metabolite levels. Alterations in neuroinflammation in the brain were examined by measuring microglial activation, inflammatory cytokine levels and the proinflammatory signaling pathway. The intestinal barrier protector intestinal alkaline phosphatase (IAP) and intestinal homeostasis inhibitor L-phenylalanine (L-Phe) were used to examine the relationship between intestinal barrier dysfunction and neuroinflammation in mice with HF. Results Eight weeks later, WT mice with HF displayed obvious increases in intestinal permeability and plasma lipopolysaccharide (LPS) levels, which were accompanied by elevated expression of TLR4 in the brain and enhanced neuroinflammation. Treatment with the intestinal barrier protector IAP significantly attenuated neuroinflammation after HF while effectively increasing plasma LPS levels. TLR4-KO mice showed significant improvements in HF-induced neuroinflammation, which was not markedly affected by intestinal barrier inhibitors or protectors. Conclusion HF could induce intestinal barrier dysfunction and increase gut-to-blood translocation of LPS, which could further promote neuroinflammation through the TLR4 pathway.
Collapse
Affiliation(s)
- Jun-Yu Huo
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wan-Ying Jiang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Yin
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai Xu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi-Ting Lyu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan-Yuan Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Geng
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Xin Jiang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi-Jun Shan
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
54
|
Lindfors S, Polianskyte-Prause Z, Bouslama R, Lehtonen E, Mannerla M, Nisen H, Tienari J, Salmenkari H, Forsgård R, Mirtti T, Lehto M, Groop PH, Lehtonen S. Adiponectin receptor agonist AdipoRon ameliorates renal inflammation in diet-induced obese mice and endotoxin-treated human glomeruli ex vivo. Diabetologia 2021; 64:1866-1879. [PMID: 33987714 PMCID: PMC8245393 DOI: 10.1007/s00125-021-05473-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/01/2021] [Indexed: 11/10/2022]
Abstract
AIMS/HYPOTHESIS Chronic low-grade inflammation with local upregulation of proinflammatory molecules plays a role in the progression of obesity-related renal injury. Reduced serum concentration of anti-inflammatory adiponectin may promote chronic inflammation. Here, we investigated the potential anti-inflammatory and renoprotective effects and mechanisms of action of AdipoRon, an adiponectin receptor agonist. METHODS Wild-type DBA/2J mice were fed with high-fat diet (HFD) supplemented or not with AdipoRon to model obesity-induced metabolic endotoxaemia and chronic low-grade inflammation and we assessed changes in the glomerular morphology and expression of proinflammatory markers. We also treated human glomeruli ex vivo and human podocytes in vitro with AdipoRon and bacterial lipopolysaccharide (LPS), an endotoxin upregulated in obesity and diabetes, and analysed the secretion of inflammatory cytokines, activation of inflammatory signal transduction pathways, apoptosis and migration. RESULTS In HFD-fed mice, AdipoRon attenuated renal inflammation, as demonstrated by reduced expression of glomerular activated NF-κB p65 subunit (NF-κB-p65) (70%, p < 0.001), TNFα (48%, p < 0.01), IL-1β (51%, p < 0.001) and TGFβ (46%, p < 0.001), renal IL-6 and IL-4 (21% and 20%, p < 0.05), and lowered glomerular F4/80-positive macrophage infiltration (31%, p < 0.001). In addition, AdipoRon ameliorated HFD-induced glomerular hypertrophy (12%, p < 0.001), fibronectin accumulation (50%, p < 0.01) and podocyte loss (12%, p < 0.001), and reduced podocyte foot process effacement (15%, p < 0.001) and thickening of the glomerular basement membrane (18%, p < 0.001). In cultured podocytes, AdipoRon attenuated the LPS-induced activation of the central inflammatory signalling pathways NF-κB-p65, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38-MAPK) (30%, 36% and 22%, respectively, p < 0.001), reduced the secretion of TNFα (32%, p < 0.01), and protected against podocyte apoptosis and migration. In human glomeruli ex vivo, AdipoRon reduced the LPS-induced secretion of inflammatory cytokines IL-1β, IL-18, IL-6 and IL-10. CONCLUSIONS/INTERPRETATION AdipoRon attenuated the renal expression of proinflammatory cytokines in HFD-fed mice and LPS-stimulated human glomeruli, which apparently contributed to the amelioration of glomerular inflammation and injury. Mechanistically, based on assays on cultured podocytes, AdipoRon reduced LPS-induced activation of the NF-κB-p65, JNK and p38-MAPK pathways, thereby impelling the decrease in apoptosis, migration and secretion of TNFα. We conclude that the activation of the adiponectin receptor by AdipoRon is a potent strategy to attenuate endotoxaemia-associated renal inflammation.
Collapse
Affiliation(s)
- Sonja Lindfors
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Zydrune Polianskyte-Prause
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Rim Bouslama
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eero Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Miia Mannerla
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harry Nisen
- Abdominal Center, Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jukka Tienari
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hanne Salmenkari
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Richard Forsgård
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuomas Mirtti
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Markku Lehto
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Per-Henrik Groop
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sanna Lehtonen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Pathology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
55
|
Larrick JW, Mendelsohn AR. Supplementation with Brush Border Enzyme Alkaline Phosphatase Slows Aging. Rejuvenation Res 2021; 23:171-175. [PMID: 32253980 DOI: 10.1089/rej.2020.2335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Diminished integrity of the intestinal epithelial barrier with advanced age is believed to contribute to aging-associated dysfunction and pathologies in animals. In mammals, diminished gut integrity contributes to inflammaging, the increase in inflammatory processes observed in old age. Recent work suggests that expression of intestinal alkaline phosphatase (IAP) plays a key role in maintaining gut integrity. IAP expression decreases with increasing age in mice and humans. Absence of IAP leads to liver inflammation and shortened life-spans in mice lacking the IAP gene. In normal mice, exogenous supplemental IAP reverses age-induced barrier dysfunction, improves aging-associated metabolic dysfunction, prevents microbiome dysbiosis (imbalance), and extends life-span. Consistent with IAP playing a conserved role in maintaining gut integrity, increased dietary IAP increases aging-diminished physical performance in flies. IAP helps maintain gut integrity in part by supporting the expression of tight junction proteins that maintain the intestinal epithelial barrier and by inactivating bacterial pro-inflammatory factors such as lipopolysaccharides (LPS) by dephosphorylation. Recombinant IAP is in late clinical trials for sepsis-associated acute kidney injury, suggesting it may soon become available as a therapeutic. Taken together, these reports support the idea that directly increasing IAP levels by supplemental recombinant IAP or by indirectly increasing IAP levels using dietary means to induce endogenous IAP may slow the development of aging-associated pathologies.
Collapse
Affiliation(s)
- James W Larrick
- Panorama Research Institute, Sunnydale, California.,Regenerative Sciences Institute, Sunnydale, California
| | - Andrew R Mendelsohn
- Panorama Research Institute, Sunnydale, California.,Regenerative Sciences Institute, Sunnydale, California
| |
Collapse
|
56
|
Association of Serum Alkaline Phosphatase with the TG/HDL Ratio and TyG Index in Korean Adults. Biomolecules 2021; 11:biom11060882. [PMID: 34198561 PMCID: PMC8231902 DOI: 10.3390/biom11060882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 11/17/2022] Open
Abstract
Alkaline phosphatase (ALP) has long been considered a marker of hepatobiliary and bone disorders, but recent studies have shown that increased ALP activity is correlated with various cardio-metabolic diseases. Thus, we investigated the association of serum ALP level with surrogate markers of insulin resistance such as triglyceride to high-density lipoprotein cholesterol ratio (TG/HDL-C ratio) and triglyceride and glucose (TyG) index in the general population. The study included 12,868 men and women aged 19 years and older. Participants were categorized into four groups based on serum ALP level (U/L) as follows: Q1: 55-190 U/L, Q2: 191-224 U/L, Q3: 225-265 U/L, and Q4: 266-923 U/L for men, Q1: 48-161 U/L, Q2: 162-198 U/L, Q3: 199-245 U/L, Q4: 246-790 U/L for women. The insulin resistance cut-off levels were defined corresponding to the 75th percentile of the TyG index and TG/HDL-C ratio in the current samples. Odds ratios (ORs) with 95% confidence intervals (CIs) of insulin resistance according to quartile of serum ALP level were calculated using weighted multivariate logistic regression analysis. Compared with Q1, the adjusted OR (95% CI) for insulin resistance of the Q4 serum ALP group was 1.517 (1.234-1.866) in men and 1.881 (1.399-2.528) in women using the TG/HDL-C ratio and 1.374 (1.093-1.728) in men and 2.047 (1.468-2.855) in women using the TyG index after adjusting for confounding variables. Serum ALP levels are independently and positively associated with surrogate markers of insulin resistance in Korean adults.
Collapse
|
57
|
Zhao C, Chen G, Wang H, Zhao Y, Chai R. Bio-inspired intestinal scavenger from microfluidic electrospray for detoxifying lipopolysaccharide. Bioact Mater 2021; 6:1653-1662. [PMID: 33313445 PMCID: PMC7701841 DOI: 10.1016/j.bioactmat.2020.11.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/28/2020] [Accepted: 11/12/2020] [Indexed: 01/03/2023] Open
Abstract
Lipopolysaccharide (LPS) plays an important role in metabolic syndrome (MetS) and other gut-derived diseases, and detoxifying LPS is considered to be a fundamental approach to prevent and treat these diseases. Here, inspired by the feeding behaviour of scavenger, novel microfluidic microcapsules with alkaline phosphatase (ALP) encapsulation and the scavenger-like molecular sieve shell are presented for cleaning intestinal LPS. Benefiting from the precisely controlled of the pore size and microfluidic electrospray, the generated microcapsules were imparted with porous molecular-sieve shells and ALP encapsulated active cores. These microcapsules could continuously work as an intestinal scavenger after colonized in intestine. It has been demonstrated that the microcapsules could englobe LPS while inhibit the permeation of digestive enzyme, and this ability contributes to promising ALP's activity, protecting cells at the presence of LPS and reducing inflammation. In addition, this scavenger inspired microcapsule could effectively decrease the LPS in organs, reduce inflammation and regulating fat metabolism in vivo. These features make the ALP encapsulated microcapsules an ideal candidate for treating MetS and other LPS related diseases.
Collapse
Affiliation(s)
- Cheng Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210002, China
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Department of Endocrinology, Shenzhen Second People's Hospital, Center for Diabetes, Obesity and Metabolic Diseases of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, 518035, PR China
| | - Guopu Chen
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210002, China
| | - Huan Wang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210002, China
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Department of Endocrinology, Shenzhen Second People's Hospital, Center for Diabetes, Obesity and Metabolic Diseases of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, 518035, PR China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210002, China
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Department of Endocrinology, Shenzhen Second People's Hospital, Center for Diabetes, Obesity and Metabolic Diseases of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, 518035, PR China
| | - Renjie Chai
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210002, China
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
58
|
Huo JY, Jiang WY, Lyu YT, Zhu L, Liu HH, Chen YY, Chen M, Geng J, Jiang ZX, Shan QJ. Renal Denervation Attenuates Neuroinflammation in the Brain by Regulating Gut-Brain Axis in Rats With Myocardial Infarction. Front Cardiovasc Med 2021; 8:650140. [PMID: 33981735 PMCID: PMC8109795 DOI: 10.3389/fcvm.2021.650140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/16/2021] [Indexed: 12/29/2022] Open
Abstract
Aims: The development of neuroinflammation deteriorates the prognosis of myocardial infarction (MI). We aimed to investigate the effect of renal denervation (RDN) on post-MI neuroinflammation in rats and the related mechanisms. Methods and Results: Male adult Sprague-Dawley rats were subjected to sham or ligation of the left anterior descending coronary artery to induce MI. One week later, the MI rats received a sham or RDN procedure. Their cardiac functions were analyzed by echocardiography, and their intestinal structures, permeability, and inflammatory cytokines were tested. The intestinal microbiota were characterized by 16S rDNA sequencing. The degrees of neuroinflammation in the brains of rats were analyzed for microglia activation, inflammatory cytokines, and inflammation-related signal pathways. In comparison with the Control rats, the MI rats exhibited impaired cardiac functions, intestinal injury, increased intestinal barrier permeability, and microbial dysbiosis, accompanied by increased microglia activation and pro-inflammatory cytokine levels in the brain. A RDN procedure dramatically decreased the levels of renal and intestinal sympathetic nerve activity, improved cardiac functions, and mitigated the MI-related intestinal injury and neuroinflammation in the brain of MI rats. Interestingly, the RDN procedure mitigated the MI-increased intestinal barrier permeability and pro-inflammatory cytokines and plasma LPS as well as ameliorated the gut microbial dysbiosis in MI rats. The protective effect of RDN was not significantly affected by treatment with intestinal alkaline phosphatase but significantly reduced by L-phenylalanine treatment in MI rats. Conclusions: RDN attenuated the neuroinflammation in the brain of MI rats, associated with mitigating the MI-related intestinal injury.
Collapse
Affiliation(s)
- Jun-Yu Huo
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wan-Ying Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi-Ting Lyu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Zhu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui-Hui Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan-Yuan Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Geng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Xin Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi-Jun Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
59
|
Wu H, Wang Y, Li H, Meng L, Zheng N, Wang J. Effect of Food Endotoxin on Infant Health. Toxins (Basel) 2021; 13:298. [PMID: 33922125 PMCID: PMC8143472 DOI: 10.3390/toxins13050298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 01/07/2023] Open
Abstract
Endotoxin is a complex molecule derived from the outer membrane of Gram-negative bacteria, and it has strong thermal stability. The processing of infant food can kill pathogenic bacteria but cannot remove endotoxin. Because the intestinal structure of infants is not fully developed, residual endotoxin poses a threat to their health by damaging the intestinal flora and inducing intestinal inflammation, obesity, and sepsis, among others. This paper discusses the sources and contents of endotoxin in infant food and methods for preventing endotoxin from harming infants. However, there is no clear evidence that endotoxin levels in infant food cause significant immune symptoms or even diseases in infants. However, in order to improve the safety level of infant food and reduce the endotoxin content, this issue should not be ignored. The purpose of this review is to provide a theoretical basis for manufacturers and consumers to understand the possible harm of endotoxin content in infant formula milk powder and to explore how to reduce its level in infant formula milk powder. Generally, producers should focus on cleaning the milk source, securing the cold chain, avoiding long-distance transportation, and shortening the storage time of raw milk to reduce the level of bacteria and endotoxin. After production and processing, the endotoxin content should be measured as an important index to test the quality of infant formula milk powder so as to provide high-quality infant products for the healthy growth of newborns.
Collapse
Affiliation(s)
- Haoming Wu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (N.Z.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China;
| | - Huiying Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (N.Z.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lu Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (N.Z.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (N.Z.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (N.Z.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
60
|
Ghosh SS, Wang J, Yannie PJ, Cooper RC, Sandhu YK, Kakiyama G, Korzun WJ, Ghosh S. Over-Expression of Intestinal Alkaline Phosphatase Attenuates Atherosclerosis. Circ Res 2021; 128:1646-1659. [PMID: 33834851 DOI: 10.1161/circresaha.120.317144] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Siddhartha S Ghosh
- Internal Medicine (S.S.G., J.W., Y.K.S., G.K., S.G.), Virginia Commonwealth University (VCU) Medical Center, Richmond, VA
| | - Jing Wang
- Internal Medicine (S.S.G., J.W., Y.K.S., G.K., S.G.), Virginia Commonwealth University (VCU) Medical Center, Richmond, VA
| | - Paul J Yannie
- Hunter Homes McGuire VA Medical Center, Richmond (P.J.Y., G.K., S.G.)
| | | | - Yashnoor K Sandhu
- Internal Medicine (S.S.G., J.W., Y.K.S., G.K., S.G.), Virginia Commonwealth University (VCU) Medical Center, Richmond, VA
| | - Genta Kakiyama
- Internal Medicine (S.S.G., J.W., Y.K.S., G.K., S.G.), Virginia Commonwealth University (VCU) Medical Center, Richmond, VA.,Hunter Homes McGuire VA Medical Center, Richmond (P.J.Y., G.K., S.G.)
| | - William J Korzun
- Clinical and Laboratory Sciences (W.J.K.), Virginia Commonwealth University (VCU) Medical Center, Richmond, VA
| | - Shobha Ghosh
- Internal Medicine (S.S.G., J.W., Y.K.S., G.K., S.G.), Virginia Commonwealth University (VCU) Medical Center, Richmond, VA.,Hunter Homes McGuire VA Medical Center, Richmond (P.J.Y., G.K., S.G.)
| |
Collapse
|
61
|
Rapid identification of magnesium ascorbyl phosphate utilizing phosphatase through a chromogenic change-coupled activity assay. Appl Microbiol Biotechnol 2021; 105:2901-2909. [PMID: 33754168 DOI: 10.1007/s00253-021-11229-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/02/2021] [Accepted: 03/10/2021] [Indexed: 10/21/2022]
Abstract
In this study, we report a chromogenic reaction between magnesium ascorbyl phosphate (MAP) and ferric chloride to generate a Brown-Red clathrate, while the Treated MAP by phosphatases forms Colorless (BRTC) product with ferric chloride. The BRTC was indicative of phosphatase activity-mediated excision of phosphorous group from MAP and utilized to screen phosphatases from bacterial cell lysates. From ten tested strains, BRTC was observed in the cell lysate of Salmonella enterica subsp. enterica serovar Cerro 87. BRTC was again employed to track phosphatase activity of the resuspensions of the ammonium sulfate graded precipitations of the cell lysate. Two phosphatases, PhoN and YcdX, were identified by LC-MS/MS analysis in the protein fraction giving most obvious BRTC phenotype and validated by examination of in vitro activity of the purified proteins. KEY POINTS: • BRTC is labelling-free, naked-eye visible, and independent of any facilities. • BRTC can directly screen phosphatases from microbial cell lysates. • Using BRTC system, two phosphatases were identified in Salmonella enterica subsp. enterica serovar Cerro 87.
Collapse
|
62
|
Wang S, Huang M, Hua J, Wei L, Lin S, Xiao L. Digital counting of single semiconducting polymer nanoparticles for the detection of alkaline phosphatase. NANOSCALE 2021; 13:4946-4955. [PMID: 33629092 DOI: 10.1039/d0nr09232k] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Alkaline phosphatase (ALP) as a necessary hydrolase in phosphate metabolism is closely related to various diseases. Ultrasensitive detection of ALP with a convenient and sensitive method is of fundamental importance. In this work, a fluorescence resonance energy transfer (FRET)-based single-particle enumeration (SPE) method is proposed for the quantitative analysis of ALP. This strategy is based on the effective fluorescence suppression by a polydopamine (PDA) shell on the surface of semiconducting polymer nanoparticles (SPNs). PDA with broadband absorption in the UV-vis region can serve as an excellent quencher for SPNs. However, ascorbic acid (AA), the product of the hydrolysis of 2-phosphate-l-ascorbic acid trisodium salt (AAP) in the presence of ALP, can effectively inhibit the self-polymerization of dopamine (DA) to form a PDA layer. Therefore, ALP can be accurately quantified by counting the concentration-related fluorescent particles in the fluorescence image. A linear range from 0.031 to 12.4 μU mL-1 and a limit-of-detection (LOD) of 0.01 μU mL-1 for ALP determination are achieved. The spiked recoveries for ALP determination in a human serum sample are between 90% and 108% with RSD less than 3.1%. In summary, this convenient and sensitive approach proposed here provides promising prospects for ALP detection in a complex biological matrix.
Collapse
Affiliation(s)
- Shumin Wang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research, Ministry of Education, Key Laboratory of Phytochemical R&D of Hunan Province, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China. and State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Mengna Huang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research, Ministry of Education, Key Laboratory of Phytochemical R&D of Hunan Province, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China.
| | - Jianhao Hua
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Lin Wei
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research, Ministry of Education, Key Laboratory of Phytochemical R&D of Hunan Province, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, China.
| | - Shen Lin
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Lehui Xiao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
63
|
Intestinal Alkaline Phosphatase Combined with Voluntary Physical Activity Alleviates Experimental Colitis in Obese Mice. Involvement of Oxidative Stress, Myokines, Adipokines and Proinflammatory Biomarkers. Antioxidants (Basel) 2021; 10:antiox10020240. [PMID: 33557311 PMCID: PMC7914798 DOI: 10.3390/antiox10020240] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/20/2021] [Accepted: 01/29/2021] [Indexed: 01/01/2023] Open
Abstract
Intestinal alkaline phosphatase (IAP) is an essential mucosal defense factor involved in the process of maintenance of gut homeostasis. We determined the effect of moderate exercise (voluntary wheel running) with or without treatment with IAP on the course of experimental murine 2,4,6-trinitrobenzenesulfonic acid (TNBS) colitis by assessing disease activity index (DAI), colonic blood flow (CBF), plasma myokine irisin levels and the colonic and adipose tissue expression of proinflammatory cytokines, markers of oxidative stress (SOD2, GPx) and adipokines in mice fed a standard diet (SD) or high-fat diet (HFD). Macroscopic and microscopic colitis in sedentary SD mice was accompanied by a significant decrease in CBF, and a significant increase in the colonic expression of tumor necrosis factor-alpha (TNF-α), IL-6, IL-1β and leptin mRNAs and decrease in the mRNA expression of adiponectin. These effects were aggravated in sedentary HFD mice but reduced in exercising animals, potentiated by concomitant treatment with IAP, especially in obese mice. Exercising HFD mice demonstrated a substantial increase in the mRNA for adiponectin and a decrease in mRNA leptin expression in intestinal mucosa and mesenteric fat as compared to sedentary animals. The expression of SOD2 and GPx mRNAs was significantly decreased in adipose tissue in HFD mice, but these effects were reversed in exercising mice with IAP administration. Our study shows for the first time that the combination of voluntary exercise and oral IAP treatment synergistically favored healing of intestinal inflammation, strengthened the antioxidant defense and ameliorated the course of experimental colitis; thus, IAP may represent a novel adjuvant therapy to alleviate inflammatory bowel disease (IBD) in humans.
Collapse
|
64
|
Pothuraju R, Chaudhary S, Rachagani S, Kaur S, Roy HK, Bouvet M, Batra SK. Mucins, gut microbiota, and postbiotics role in colorectal cancer. Gut Microbes 2021; 13:1974795. [PMID: 34586012 PMCID: PMC8489937 DOI: 10.1080/19490976.2021.1974795] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
An imbalance in the crosstalk between the host and gut microbiota affects the intestinal barrier function, which results in inflammatory diseases and colorectal cancer. The colon epithelium protects itself from a harsh environment and various pathogenic organisms by forming a double mucus layer, primarily comprising mucins. Recent studies are focusing on how dietary patterns alter the gut microbiota composition, which in turn regulates mucin expression and maintains the intestinal layers. In addition, modulation of gut microbiota by microbiotic therapy (involving fecal microbiota transplantation) has emerged as a significant factor in the pathologies associated with dysbiosis. Therefore, proper communication between host and gut microbiota via different dietary patterns (prebiotics and probiotics) is needed to maintain mucus composition, mucin synthesis, and regulation. Here, we review how the interactions between diet and gut microbiota and bacterial metabolites (postbiotics) regulate mucus layer functionalities and mucin expression in human health and disease.
Collapse
Affiliation(s)
- Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjib Chaudhary
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hemant K. Roy
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Michael Bouvet
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
65
|
Liu Y, Cavallaro PM, Kim BM, Liu T, Wang H, Kühn F, Adiliaghdam F, Liu E, Vasan R, Samarbafzadeh E, Farber MZ, Li J, Xu M, Mohad V, Choi M, Hodin RA. A role for intestinal alkaline phosphatase in preventing liver fibrosis. Theranostics 2021; 11:14-26. [PMID: 33391458 PMCID: PMC7681079 DOI: 10.7150/thno.48468] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Rationale: Liver fibrosis is frequently associated with gut barrier dysfunction, and the lipopolysaccharides (LPS) -TLR4 pathway is common to the development of both. Intestinal alkaline phosphatase (IAP) has the ability to detoxify LPS, as well as maintain intestinal tight junction proteins and gut barrier integrity. Therefore, we hypothesized that IAP may function as a novel therapy to prevent liver fibrosis. Methods: Stool IAP activity from cirrhotic patients were determined. Common bile duct ligation (CBDL) and Carbon Tetrachloride-4 (CCl4)-induced liver fibrosis models were used in WT, IAP knockout (KO), and TLR4 KO mice supplemented with or without exogenous IAP in their drinking water. The gut barrier function and liver fibrosis markers were tested. Results: Human stool IAP activity was decreased in the setting of liver cirrhosis. In mice, IAP activity and genes expression decreased after CBDL and CCl4 exposure. Intestinal tight junction related genes and gut barrier function were impaired in both models of liver fibrosis. Oral IAP supplementation attenuated the decrease in small intestine tight junction protein gene expression and gut barrier function. Liver fibrosis markers were significantly higher in IAP KO compared to WT mice in both models, while oral IAP rescued liver fibrosis in both WT and IAP KO mice. In contrast, IAP supplementation did not attenuate fibrosis in TLR4 KO mice in either model. Conclusions: Endogenous IAP is decreased during liver fibrosis, perhaps contributing to the gut barrier dysfunction and worsening fibrosis. Oral IAP protects the gut barrier and further prevents the development of liver fibrosis via a TLR4-mediated mechanism.
Collapse
Affiliation(s)
- Yang Liu
- Department of General Surgery, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, China
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Paul M. Cavallaro
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Byeong-Moo Kim
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Tao Liu
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
- Department of Gastroenterological Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyan Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
- Department of Gastroenterological Surgery, People's Hospital of Liaoning Province, Shenyang, China
| | - Florian Kühn
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Enyu Liu
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jian, China
| | - Robin Vasan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
- Department of Surgery, University-Pittsburgh Medical Center, Pittsburgh, PA, US
| | - Ehsan Samarbafzadeh
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Matthew Z. Farber
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Junhui Li
- Department of General Surgery, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, China
| | - Meng Xu
- Department of General Surgery, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an, China
| | - Vidisha Mohad
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Michael Choi
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Richard A. Hodin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| |
Collapse
|
66
|
Gámez-Belmonte R, Tena-Garitaonaindia M, Hernández-Chirlaque C, Córdova S, Ceacero-Heras D, de Medina FS, Martínez-Augustin O. Deficiency in Tissue Non-Specific Alkaline Phosphatase Leads to Steatohepatitis in Mice Fed a High Fat Diet Similar to That Produced by a Methionine and Choline Deficient Diet. Int J Mol Sci 2020; 22:ijms22010051. [PMID: 33374541 PMCID: PMC7793076 DOI: 10.3390/ijms22010051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 01/15/2023] Open
Abstract
The liver expresses tissue-nonspecific alkaline phosphatase (TNAP), which may participate in the defense against bacterial components, in cell regulation as part of the purinome or in bile secretion, among other roles. We aimed to study the role of TNAP in the development of hepatosteatosis. TNAP+/− haplodeficient and wild type (WT) mice were fed a control diet (containing 10% fat w/w) or the same diet deficient in methionine and choline (MCD diet). The MCD diet induced substantial weight loss together with hepatic steatosis and increased alanine aminotransferase (ALT) plasma levels, but no differences in IL-6, TNF, insulin or resistin. There were no substantial differences between TNAP+/− and WT mice fed the MCD diet. In turn, TNAP+/− mice receiving the control diet presented hepatic steatosis with alterations in metabolic parameters very similar to those induced by the MCD diet. Nevertheless, no weight loss, increased ALT plasma levels or hypoglycemia were observed. These mice also presented increased levels of liver TNF and systemic resistin and glucagon compared to WT mice. The phenotype of TNAP+/− mice fed a standard diet was normal. In conclusion, TNAP haplodeficiency induces steatosis comparable to that produced by a MCD diet when fed a control diet.
Collapse
Affiliation(s)
- Reyes Gámez-Belmonte
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain;
| | - Mireia Tena-Garitaonaindia
- Department of Biochemistry and Molecular Biology 2, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, Instituto de Nutrición y Tecnología de los Alimentos José Mataix, University of Granada, 18071 Granada, Spain; (M.T.-G.); (C.H.-C.); (S.C.); (D.C.-H.); (O.M.-A.)
| | - Cristina Hernández-Chirlaque
- Department of Biochemistry and Molecular Biology 2, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, Instituto de Nutrición y Tecnología de los Alimentos José Mataix, University of Granada, 18071 Granada, Spain; (M.T.-G.); (C.H.-C.); (S.C.); (D.C.-H.); (O.M.-A.)
| | - Samir Córdova
- Department of Biochemistry and Molecular Biology 2, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, Instituto de Nutrición y Tecnología de los Alimentos José Mataix, University of Granada, 18071 Granada, Spain; (M.T.-G.); (C.H.-C.); (S.C.); (D.C.-H.); (O.M.-A.)
| | - Diego Ceacero-Heras
- Department of Biochemistry and Molecular Biology 2, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, Instituto de Nutrición y Tecnología de los Alimentos José Mataix, University of Granada, 18071 Granada, Spain; (M.T.-G.); (C.H.-C.); (S.C.); (D.C.-H.); (O.M.-A.)
| | - Fermín Sánchez de Medina
- Department of Pharmacology, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, 18071 Granada, Spain;
- Correspondence: ; Tel.: +34-958-241747
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology 2, CIBERehd, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, Instituto de Nutrición y Tecnología de los Alimentos José Mataix, University of Granada, 18071 Granada, Spain; (M.T.-G.); (C.H.-C.); (S.C.); (D.C.-H.); (O.M.-A.)
| |
Collapse
|
67
|
Munford RS, Weiss JP, Lu M. Biochemical transformation of bacterial lipopolysaccharides by acyloxyacyl hydrolase reduces host injury and promotes recovery. J Biol Chem 2020; 295:17842-17851. [PMID: 33454018 DOI: 10.1074/jbc.rev120.015254] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/22/2020] [Indexed: 12/26/2022] Open
Abstract
Animals can sense the presence of microbes in their tissues and mobilize their own defenses by recognizing and responding to conserved microbial structures (often called microbe-associated molecular patterns (MAMPs)). Successful host defenses may kill the invaders, yet the host animal may fail to restore homeostasis if the stimulatory microbial structures are not silenced. Although mice have many mechanisms for limiting their responses to lipopolysaccharide (LPS), a major Gram-negative bacterial MAMP, a highly conserved host lipase is required to extinguish LPS sensing in tissues and restore homeostasis. We review recent progress in understanding how this enzyme, acyloxyacyl hydrolase (AOAH), transforms LPS from stimulus to inhibitor, reduces tissue injury and death from infection, prevents prolonged post-infection immunosuppression, and keeps stimulatory LPS from entering the bloodstream. We also discuss how AOAH may increase sensitivity to pulmonary allergens. Better appreciation of how host enzymes modify LPS and other MAMPs may help prevent tissue injury and hasten recovery from infection.
Collapse
Affiliation(s)
- Robert S Munford
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, Maryland, USA.
| | - Jerrold P Weiss
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA
| | - Mingfang Lu
- Department of Immunology and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
68
|
Schippers M, Post E, Eichhorn I, Langeland J, Beljaars L, Malo MS, Hodin RA, Millán JL, Popov Y, Schuppan D, Poelstra K. Phosphate Groups in the Lipid A Moiety Determine the Effects of LPS on Hepatic Stellate Cells: A Role for LPS-Dephosphorylating Activity in Liver Fibrosis. Cells 2020; 9:E2708. [PMID: 33348845 PMCID: PMC7766276 DOI: 10.3390/cells9122708] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022] Open
Abstract
Alkaline phosphatase (AP) activity is highly upregulated in plasma during liver diseases. Previously, we demonstrated that AP is able to detoxify lipopolysaccharide (LPS) by dephosphorylating its lipid A moiety. Because a role of gut-derived LPS in liver fibrogenesis has become evident, we now examined the relevance of phosphate groups in the lipid A moiety in this process. The effects of mono-phosphoryl and di-phosphoryl lipid A (MPLA and DPLA, respectively) were studied in vitro and LPS-dephosphorylating activity was studied in normal and fibrotic mouse and human livers. The effects of intestinal AP were studied in mice with CCL4-induced liver fibrosis. DPLA strongly stimulated fibrogenic and inflammatory activities in primary rat hepatic stellate cells (rHSCs) and RAW264.7 macrophages with similar potency as full length LPS. However, MPLA did not affect any of the parameters. LPS-dephosphorylating activity was found in mouse and human livers and was strongly increased during fibrogenesis. Treatment of fibrotic mice with intravenous intestinal-AP significantly attenuated intrahepatic desmin+- and αSMA+ -HSC and CD68+- macrophage accumulation. In conclusion, the lack of biological activity of MPLA, contrasting with the profound activities of DPLA, shows the relevance of LPS-dephosphorylating activity. The upregulation of LPS-dephosphorylating activity in fibrotic livers and the protective effects of exogenous AP during fibrogenesis indicate an important physiological role of intestinal-derived AP during liver fibrosis.
Collapse
Affiliation(s)
- Marlies Schippers
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Eduard Post
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Ilse Eichhorn
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Jitske Langeland
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Leonie Beljaars
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| | - Madhu S. Malo
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (M.S.M.); (R.A.H.)
- Bangladesh Institute of Research and Rehabilitation for Diabetes, Endocrine and Metabolic Disorders (BIRDEM), Dhaka 1000, Bangladesh
| | - Richard A. Hodin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (M.S.M.); (R.A.H.)
| | - José Luis Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
| | - Yury Popov
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (Y.P.); (D.S.)
| | - Detlef Schuppan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (Y.P.); (D.S.)
- Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Klaas Poelstra
- Department of Nanomedice and Drug Targeting, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (M.S.); (E.P.); (I.E.); (J.L.); (L.B.)
| |
Collapse
|
69
|
Alvarenga L, Cardozo LFMF, Lindholm B, Stenvinkel P, Mafra D. Intestinal alkaline phosphatase modulation by food components: predictive, preventive, and personalized strategies for novel treatment options in chronic kidney disease. EPMA J 2020; 11:565-579. [PMID: 33240450 PMCID: PMC7680467 DOI: 10.1007/s13167-020-00228-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/30/2020] [Indexed: 12/18/2022]
Abstract
Alkaline phosphatase (AP) is a ubiquitous membrane-bound glycoprotein that catalyzes phosphate monoesters' hydrolysis from organic compounds, an essential process in cell signaling. Four AP isozymes have been described in humans, placental AP, germ cell AP, tissue nonspecific AP, and intestinal AP (IAP). IAP plays a crucial role in gut microbial homeostasis, nutrient uptake, and local and systemic inflammation, and its dysfunction is associated with persistent inflammatory disorders. AP is a strong predictor of mortality in the general population and patients with cardiovascular and chronic kidney disease (CKD). However, little is known about IAP modulation and its possible consequences in CKD, a disease characterized by gut microbiota imbalance and persistent low-grade inflammation. Mitigating inflammation and dysbiosis can prevent cardiovascular complications in patients with CKD, and monitoring factors such as IAP can be useful for predicting those complications. Here, we review IAP's role and the results of nutritional interventions targeting IAP in experimental models to prevent alterations in the gut microbiota, which could be a possible target of predictive, preventive, personalized medicine (PPPM) to avoid CKD complications. Microbiota and some nutrients may activate IAP, which seems to have a beneficial impact on health; however, data on CKD remains scarce.
Collapse
Affiliation(s)
- L. Alvarenga
- Post Graduation Program in Medical Sciences, (UFF) Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói, Brazil
| | - L. F. M. F. Cardozo
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói, Rio de Janeiro (RJ) Brazil
| | - B. Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - P. Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - D. Mafra
- Post Graduation Program in Medical Sciences, (UFF) Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói, Brazil
- Post Graduation Program in Cardiovascular Sciences, Federal Fluminense University (UFF), Niterói, Rio de Janeiro (RJ) Brazil
| |
Collapse
|
70
|
Abstract
PURPOSE OF REVIEW The gut barrier serves as the primary interface between the environment and host in terms of surface area and complexity. Luminal chemosensing is a term used to describe how small molecules in the gut lumen interact with the host through surface receptors or via transport into the subepithelial space. In this review, we have summarized recent advances in the understanding of the luminal chemosensory system in the gastroduodenal epithelium consisting of enterocytes, enteroendocrine, and tuft cells, with particular emphasis on how chemosensing affects mucosal protective responses and the metabolic syndrome. RECENT FINDINGS Recent single-cell RNA sequencing provides detailed cell type-specific expression of chemosensory receptors and other bioactive molecules as well as cell lineages; some are similar to lingual taste cells whereas some are gut specific. Gut luminal chemosensing is not only important for the local or remote regulation of gut function, but also contributes to the systemic regulation of metabolism, energy balance, and food intake. We will discuss the chemosensory mechanisms of the proximal intestine, in particular to gastric acid, with a focus on the cell types and receptors involved in chemosensing, with emphasis on the rare chemosensory cells termed tuft cells. We will also discuss the chemosensory functions of intestinal ectoenzymes and bacterial components (e.g., lipopolysaccharide) as well as how they affect mucosal function through altering the gut-hormonal-neural axis. SUMMARY Recent updates in luminal chemosensing by different chemosensory cells have provided new possibilities for identifying novel molecular targets for the treatment of mucosal injury, metabolic disorders, and abnormal visceral sensation.
Collapse
|
71
|
Ásgeirsson B, Markússon S, Hlynsdóttir SS, Helland R, Hjörleifsson JG. X-ray crystal structure of Vibrio alkaline phosphatase with the non-competitive inhibitor cyclohexylamine. Biochem Biophys Rep 2020; 24:100830. [PMID: 33102813 PMCID: PMC7569297 DOI: 10.1016/j.bbrep.2020.100830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 10/06/2020] [Indexed: 11/13/2022] Open
Abstract
Background Para-nitrophenyl phosphate, the common substrate for alkaline phosphatase (AP), is available as a cyclohexylamine salt. Here, we report that cyclohexylamine is a non-competitive inhibitor of APs. Methods Cyclohexylamine inhibited four different APs. Co-crystallization with the cold-active Vibrio AP (VAP) was performed and the structure solved. Results Inhibition of VAP fitted a non-competitive kinetic model (Km unchanged, Vmax reduced) with IC50 45.3 mM at the pH optimum 9.8, not sensitive to 0.5 M NaCl, and IC50 27.9 mM at pH 8.0, where the addition of 0.5 M NaCl altered the inhibition to the level observed at pH 9.8. APs from E. coli and calf intestines were less sensitive to cyclohexylamine, whereas an Antarctic bacterial AP was similar to VAP in this respect. X-ray crystallography at 2.3 Å showed two binding sites, one in the active site channel and another at the surface close to dimer interface. Antarctic bacterial AP and VAP have Trp274 in common in their active-sites, that takes part in binding cyclohexylamine. VAP variants W274A, W274K, and W274H gave IC50 values of 179 mM, 188 mM and 187 mM, respectively, at pH 9.8. Conclusions The binding of cyclohexylamine in locations at the dimeric interface and/or in the active site of APs may delay product release or reduce the rate of catalytic step(s) involving conformational changes and intersubunit communications. General significance Cyclohexylamine is a common chemical in industries and used as a counterion in substrates for alkaline phosphatase, a clinically important and common enzyme in the biosphere. Cyclohexylamine inhibits alkaline phosphatase activity non-competitively. X-ray structure was solved that shows cyclohexylamine bound to alkaline phosphatase at two sites. Alkaline phosphatases from four different organisms bind cyclohexylamine with varying affinity.
Collapse
Affiliation(s)
- Bjarni Ásgeirsson
- Department of Biochemistry, Science Institute, University of Iceland, Dunhagi 3, 107 Reykjavik, Iceland
| | - Sigurbjörn Markússon
- Department of Biochemistry, Science Institute, University of Iceland, Dunhagi 3, 107 Reykjavik, Iceland
| | - Sigríður S Hlynsdóttir
- Department of Biochemistry, Science Institute, University of Iceland, Dunhagi 3, 107 Reykjavik, Iceland
| | - Ronny Helland
- NorStruct, Department of Chemistry, Faculty of Science and Technology, UiT - the Arctic University of Norway, NO-9037, Tromsø, Norway
| | - Jens G Hjörleifsson
- Department of Biochemistry, Science Institute, University of Iceland, Dunhagi 3, 107 Reykjavik, Iceland
| |
Collapse
|
72
|
Ciesielska A, Matyjek M, Kwiatkowska K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell Mol Life Sci 2020; 78:1233-1261. [PMID: 33057840 PMCID: PMC7904555 DOI: 10.1007/s00018-020-03656-y] [Citation(s) in RCA: 797] [Impact Index Per Article: 159.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/25/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Toll-like receptor (TLR) 4 belongs to the TLR family of receptors inducing pro-inflammatory responses to invading pathogens. TLR4 is activated by lipopolysaccharide (LPS, endotoxin) of Gram-negative bacteria and sequentially triggers two signaling cascades: the first one involving TIRAP and MyD88 adaptor proteins is induced in the plasma membrane, whereas the second engaging adaptor proteins TRAM and TRIF begins in early endosomes after endocytosis of the receptor. The LPS-induced internalization of TLR4 and hence also the activation of the TRIF-dependent pathway is governed by a GPI-anchored protein, CD14. The endocytosis of TLR4 terminates the MyD88-dependent signaling, while the following endosome maturation and lysosomal degradation of TLR4 determine the duration and magnitude of the TRIF-dependent one. Alternatively, TLR4 may return to the plasma membrane, which process is still poorly understood. Therefore, the course of the LPS-induced pro-inflammatory responses depends strictly on the rates of TLR4 endocytosis and trafficking through the endo-lysosomal compartment. Notably, prolonged activation of TLR4 is linked with several hereditary human diseases, neurodegeneration and also with autoimmune diseases and cancer. Recent studies have provided ample data on the role of diverse proteins regulating the functions of early, late, and recycling endosomes in the TLR4-induced inflammation caused by LPS or phagocytosis of E. coli. In this review, we focus on the mechanisms of the internalization and intracellular trafficking of TLR4 and CD14, and also of LPS, in immune cells and discuss how dysregulation of the endo-lysosomal compartment contributes to the development of diverse human diseases.
Collapse
Affiliation(s)
- Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland.
| | - Marta Matyjek
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| |
Collapse
|
73
|
Adiliaghdam F, Cavallaro P, Mohad V, Almpani M, Kühn F, Gharedaghi MH, Najibi M, Rahme LG, Hodin RA. Targeting the gut to prevent sepsis from a cutaneous burn. JCI Insight 2020; 5:137128. [PMID: 33004693 PMCID: PMC7566703 DOI: 10.1172/jci.insight.137128] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Severe burn injury induces gut barrier dysfunction and subsequently a profound systemic inflammatory response. In the present study, we examined the role of the small intestinal brush border enzyme, intestinal alkaline phosphatase (IAP), in preserving gut barrier function and preventing systemic inflammation after burn wound infection in mice. Mice were subjected to a 30% total body surface area dorsal burn with or without intradermal injection of Pseudomonas aeruginosa. Mice were gavaged with 2000 units of IAP or vehicle at 3 and 12 hours after the insult. We found that both endogenously produced and exogenously supplemented IAP significantly reduced gut barrier damage, decreased bacterial translocation to the systemic organs, attenuated systemic inflammation, and improved survival in this burn wound infection model. IAP attenuated liver inflammation and reduced the proinflammatory characteristics of portal serum. Furthermore, we found that intestinal luminal contents of burn wound-infected mice negatively impacted the intestinal epithelial integrity compared with luminal contents of control mice and that IAP supplementation preserved monolayer integrity. These results indicate that oral IAP therapy may represent an approach to preserving gut barrier function, blocking proinflammatory triggers from entering the portal system, preventing gut-induced systemic inflammation, and improving survival after severe burn injuries.
Collapse
Affiliation(s)
- Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul Cavallaro
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vidisha Mohad
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianna Almpani
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Florian Kühn
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of General, Visceral and Transplant Surgery, Hospital of the University of Munich, Munich, Germany
| | - Mohammad Hadi Gharedaghi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mehran Najibi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Laurence G. Rahme
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard A. Hodin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
74
|
IIAEK Targets Intestinal Alkaline Phosphatase (IAP) to Improve Cholesterol Metabolism with a Specific Activation of IAP and Downregulation of ABCA1. Nutrients 2020; 12:nu12092859. [PMID: 32961978 PMCID: PMC7551322 DOI: 10.3390/nu12092859] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/10/2020] [Accepted: 09/16/2020] [Indexed: 01/11/2023] Open
Abstract
IIAEK (Ile-Ile-Ala-Glu-Lys, lactostatin) is a novel cholesterol-lowering pentapeptide derived from bovine milk β-lactoglobulin. However, the molecular mechanisms underlying the IIAEK-mediated suppression of intestinal cholesterol absorption are unknown. Therefore, we evaluated the effects of IIAEK on intestinal cholesterol metabolism in a human intestinal model using Caco-2 cells. We found that IIAEK significantly reduced the expression of intestinal cholesterol metabolism-associated genes, particularly that of the ATP-binding cassette transporter A1 (ABCA1). Subsequently, we chemically synthesized a novel molecular probe, IIXEK, which can visualize a complex of target proteins interacting with photoaffinity-labeled IIAEK by fluorescent substances. Through photoaffinity labeling and MS analysis with IIXEK for the rat small intestinal mucosa and intestinal lipid raft fractions of Caco-2 cells, we identified intestinal alkaline phosphatase (IAP) as a specific molecule interacting with IIAEK and discovered the common IIAEK-binding amino acid sequence, GFYLFVEGGR. IIAEK significantly increased IAP mRNA and protein levels while decreasing ABCA1 mRNA and protein levels in Caco-2 cells. In conclusion, we found that IIAEK targets IAP to improve cholesterol metabolism via a novel signaling pathway involving the specific activation of IAP and downregulation of intestinal ABCA1.
Collapse
|
75
|
Wong SK, Chin KY, Ima-Nirwana S. Toll-like Receptor as a Molecular Link between Metabolic Syndrome and Inflammation: A Review. Curr Drug Targets 2020; 20:1264-1280. [PMID: 30961493 DOI: 10.2174/1389450120666190405172524] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
Metabolic Syndrome (MetS) involves a cluster of five conditions, i.e. obesity, hyperglycaemia, hypertension, hypertriglyceridemia and low High-Density Lipoprotein (HDL) cholesterol. All components of MetS share an underlying chronic inflammatory aetiology, manifested by increased levels of pro-inflammatory cytokines. The pathogenic role of inflammation in the development of MetS suggested that toll-like receptor (TLR) activation may trigger MetS. This review summarises the supporting evidence on the interactions between MetS and TLR activation, bridged by the elevation of TLR ligands during MetS. The regulatory circuits mediated by TLR activation, which modulates signal propagation, leading to the state of chronic inflammation, are also discussed. Taken together, TLR activation could be the molecular basis in the development of MetS-induced inflammation.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Soelaiman Ima-Nirwana
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
76
|
Understanding the enzymatic inhibition of intestinal alkaline phosphatase by aminophenazone-derived aryl thioureas with aided computational molecular dynamics simulations: synthesis, characterization, SAR and kinetic profiling. Mol Divers 2020; 25:1701-1715. [PMID: 32862361 DOI: 10.1007/s11030-020-10136-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 08/19/2020] [Indexed: 10/23/2022]
Abstract
The work presented in this paper aims toward the synthesis of aryl thiourea derivatives 4a-l of pyrazole based nonsteroidal anti-inflammatory drug named 4-aminophenazone, as potential inhibitors of intestinal alkaline phosphatase enzyme. The screening of synthesized target compounds 4a-l for unraveling the anti-inflammatory potential against calf intestinal alkaline phosphatase gives rise to lead member 4c possessing IC50 value 0.420 ± 0.012 µM, many folds better than reference standard used (KH2PO4 IC50 = 2.8 ± 0.06 µM and L-phenylalanine IC50 = 100 ± 3.1 µM). SAR for unfolding the active site binding pocket interaction along with the mode of enzyme inhibition based on kinetic studies is carried out which showed non-competitive binding mode. The enzyme inhibition studies were further supplemented by molecular dynamic simulations for predicting the protein behavior against active inhibitors 4c and 4g during docking analysis. The preliminary toxicity of the synthesized compounds was determined by using brine shrimp assay. This work also includes detailed biochemical analysis along with RO5 parameters for all the newly synthesized drug derivatives 4a-l.
Collapse
|
77
|
Furlan Freguia C, Marriott A, Gill D, Kaleko M. Maternal treatment with oral intestinal alkaline phosphatase mitigates high fat diet-induced cognitive disorders in offspring mice. Behav Brain Res 2020; 392:112701. [PMID: 32464122 DOI: 10.1016/j.bbr.2020.112701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/09/2020] [Accepted: 05/08/2020] [Indexed: 12/28/2022]
Abstract
Intestinal alkaline phosphatase (IAP) is an endogenous enzyme that promotes gastrointestinal homeostasis by detoxifying inflammatory mediators, tightening the gut barrier and promoting a healthy microbiome. Oral IAP administration was efficacious in ameliorating diabetes in a high fat diet (HFD)-induced murine model. In humans, maternal obesity and diabetes during pregnancy have been associated with an increased risk of autism spectrum disorders (ASD). In mice, HFD-induced maternal obesity leads to offspring with cognitive deficiency. Here we investigated whether IAP administration to obese dams could ameliorate autism-like disorders in mice. Using a HFD murine model, we recapitulated that maternal obesity leads to male offspring with social deficits as shown by the three chamber test and reciprocal social interaction analyses. Notably, oral delivery of IAP to dams improved those deficiencies. In addition, a jumping behavior was noted in pups from obese dams, which was rescued by maternal IAP treatment. Our findings suggest that maternal treatment with IAP can relieve some ASD-like symptoms in offspring mice.
Collapse
|
78
|
Vitamin B6 cofactors conjugated ovalbumin-stabilized gold nanoclusters: Application in alkaline phosphatase activity detection and generating white-light emission. Microchem J 2020. [DOI: 10.1016/j.microc.2020.104859] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
79
|
Akiba Y, Maruta K, Takajo T, Narimatsu K, Said H, Kato I, Kuwahara A, Kaunitz JD. Lipopolysaccharides transport during fat absorption in rodent small intestine. Am J Physiol Gastrointest Liver Physiol 2020; 318:G1070-G1087. [PMID: 32390462 PMCID: PMC7311662 DOI: 10.1152/ajpgi.00079.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/29/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023]
Abstract
Lipopolysaccharides (LPS) are potent pro-inflammatory molecules that enter the systemic circulation from the intestinal lumen by uncertain mechanisms. We investigated these mechanisms and the effect of exogenous glucagon-like peptide-2 (GLP-2) on LPS transport in the rodent small intestine. Transmucosal LPS transport was measured in Ussing-chambered rat jejunal mucosa. In anesthetized rats, the appearance of fluorescein isothiocyanate (FITC)-LPS into the portal vein (PV) and the mesenteric lymph was simultaneously monitored after intraduodenal perfusion of FITC-LPS with oleic acid and taurocholate (OA/TCA). In vitro, luminally applied LPS rapidly appeared in the serosal solution only with luminal OA/TCA present, inhibited by the lipid raft inhibitor methyl-β-cyclodextrin (MβCD) and the CD36 inhibitor sulfosuccinimidyl oleate (SSO), or by serosal GLP-2. In vivo, perfusion of FITC-LPS with OA/TCA rapidly increased FITC-LPS appearance into the PV, followed by a gradual increase of FITC-LPS into the lymph. Rapid PV transport was inhibited by the addition of MβCD or by SSO, whereas transport into the lymph was inhibited by chylomicron synthesis inhibition. Intraveous injection of the stable GLP-2 analog teduglutide acutely inhibited FITC-LPS transport into the PV, yet accelerated FITC-LPS transport into the lymph via Nω-nitro-l-arginine methyl ester (l-NAME)- and PG97-269-sensitive mechanisms. In vivo confocal microscopy in mouse jejunum confirmed intracellular FITC-LPS uptake with no evidence of paracellular localization. This is the first direct demonstration in vivo that luminal LPS may cross the small intestinal barrier physiologically during fat absorption via lipid raft- and CD36-mediated mechanisms, followed by predominant transport into the PV, and that teduglutide inhibits LPS uptake into the PV in vivo.NEW & NOTEWORTHY We report direct in vivo confirmation of transcellular lipopolysaccharides (LPS) uptake from the intestine into the portal vein (PV) involving CD36 and lipid rafts, with minor uptake via the canonical chylomicron pathway. The gut hormone glucagon-like peptide-2 (GLP-2) inhibited uptake into the PV. These data suggest that the bulk of LPS absorption is via the PV to the liver, helping clarify the mechanism of LPS transport into the PV as part of the "gut-liver" axis. These data do not support the paracellular transport of LPS, which has been implicated in the pathogenesis of the "leaky gut" syndrome.
Collapse
Affiliation(s)
- Yasutada Akiba
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
- Brentwood Biomedical Research Institute, Los Angeles, California
| | - Koji Maruta
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
| | - Takeshi Takajo
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
| | - Kazuyuki Narimatsu
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
| | - Hyder Said
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
| | - Ikuo Kato
- Department of Medical Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Atsukazu Kuwahara
- Research Unit for Epithelial Physiology, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Japan
| | - Jonathan D Kaunitz
- Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California
- Department of Medicine, University of California, School of Medicine, Los Angeles, California
- Department of Surgery, University of California, School of Medicine, Los Angeles, California
- Brentwood Biomedical Research Institute, Los Angeles, California
| |
Collapse
|
80
|
|
81
|
Kawada T. Serum alkaline phosphatase level and metabolic syndrome. Clin Chim Acta 2020; 506:187. [PMID: 32243983 DOI: 10.1016/j.cca.2020.03.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/29/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Tomoyuki Kawada
- Department of Hygiene and Public Health, Nippon Medical School, Japan.
| |
Collapse
|
82
|
Kühn F, Adiliaghdam F, Cavallaro PM, Hamarneh SR, Tsurumi A, Hoda RS, Munoz AR, Dhole Y, Ramirez JM, Liu E, Vasan R, Liu Y, Samarbafzadeh E, Nunez RA, Farber MZ, Chopra V, Malo MS, Rahme LG, Hodin RA. Intestinal alkaline phosphatase targets the gut barrier to prevent aging. JCI Insight 2020; 5:134049. [PMID: 32213701 PMCID: PMC7213802 DOI: 10.1172/jci.insight.134049] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/20/2020] [Indexed: 12/16/2022] Open
Abstract
Gut barrier dysfunction and gut-derived chronic inflammation play crucial roles in human aging. The gut brush border enzyme intestinal alkaline phosphatase (IAP) functions to inhibit inflammatory mediators and also appears to be an important positive regulator of gut barrier function and microbial homeostasis. We hypothesized that this enzyme could play a critical role in regulating the aging process. We tested the role of several IAP functions for prevention of age-dependent alterations in intestinal homeostasis by employing different loss-of-function and supplementation approaches. In mice, there is an age-related increase in gut permeability that is accompanied by increases in gut-derived portal venous and systemic inflammation. All these phenotypes were significantly more pronounced in IAP-deficient animals. Oral IAP supplementation significantly decreased age-related gut permeability and gut-derived systemic inflammation, resulted in less frailty, and extended lifespan. Furthermore, IAP supplementation was associated with preserving the homeostasis of gut microbiota during aging. These effects of IAP were also evident in a second model system, Drosophilae melanogaster. IAP appears to preserve intestinal homeostasis in aging by targeting crucial intestinal alterations, including gut barrier dysfunction, dysbiosis, and endotoxemia. Oral IAP supplementation may represent a novel therapy to counteract the chronic inflammatory state leading to frailty and age-related diseases in humans.
Collapse
Affiliation(s)
- Florian Kühn
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
- Department of General, Visceral and Transplant Surgery, Hospital of the University of Munich, Munich, Germany
| | - Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Paul M. Cavallaro
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Sulaiman R. Hamarneh
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Amy Tsurumi
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | | | - Alexander R. Munoz
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Yashoda Dhole
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Juan M. Ramirez
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Enyu Liu
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Robin Vasan
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Yang Liu
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Ehsan Samarbafzadeh
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Rocio A. Nunez
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew Z. Farber
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Vanita Chopra
- Department of Neurology,, MGH, Harvard Medical School, Boston, Massachusetts, USA
| | - Madhu S. Malo
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Laurence G. Rahme
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Richard A. Hodin
- Department of Surgery, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
83
|
Lallès JP. Recent advances in intestinal alkaline phosphatase, inflammation, and nutrition. Nutr Rev 2020; 77:710-724. [PMID: 31086953 DOI: 10.1093/nutrit/nuz015] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In recent years, much new data on intestinal alkaline phosphatase (IAP) have been published, and major breakthroughs have been disclosed. The aim of the present review is to critically analyze the publications released over the last 5 years. These breakthroughs include, for example, the direct implication of IAP in intestinal tight junction integrity and barrier function maintenance; chronic intestinal challenge with low concentrations of Salmonella generating long-lasting depletion of IAP and increased susceptibility to inflammation; the suggestion that genetic mutations in the IAP gene in humans contribute to some forms of chronic inflammatory diseases and loss of functional IAP along the gut and in stools; stool IAP as an early biomarker of incipient diabetes in humans; and omega-3 fatty acids as direct inducers of IAP in intestinal tissue. Many recent papers have also explored the prophylactic and therapeutic potential of IAP and other alkaline phosphatase (AP) isoforms in various experimental settings and diseases. Remarkably, nearly all data confirm the potent anti-inflammatory properties of (I)AP and the negative consequences of its inhibition on health. A simplified model of the body AP system integrating the IAP compartment is provided. Finally, the list of nutrients and food components stimulating IAP has continued to grow, thus emphasizing nutrition as a potent lever for limiting inflammation.
Collapse
Affiliation(s)
- Jean-Paul Lallès
- Institut National de la Recherche Agronomique (INRA), Human Nutrition Division, Clermont-Ferrand, France, and the Centre de Recherche en Nutrition Humaine Ouest, Nantes, France
| |
Collapse
|
84
|
Yang T, Li H, Oliveira AC, Goel R, Richards EM, Pepine CJ, Raizada MK. Transcriptomic signature of gut microbiome-contacting cells in colon of spontaneously hypertensive rats. Physiol Genomics 2020; 52:121-132. [PMID: 31869283 PMCID: PMC7099409 DOI: 10.1152/physiolgenomics.00087.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 01/14/2023] Open
Abstract
Fecal matter transfer from hypertensive patients and animals into normotensive animals increases blood pressure, strengthening the evidence for gut-microbiota interactions in the control of blood pressure. However, cellular and molecular events involved in gut dysbiosis-associated hypertension remain poorly understood. Therefore, our objective in this study was to use gene expression profiling to characterize the gut epithelium layer in the colon in hypertension. We observed significant suppression of components of T cell receptor (TCR) signaling in the colonic epithelium of spontaneously hypertensive rats (SHR) when compared with Wistar Kyoto (WKY) normotensive rats. Western blot analysis confirmed lower expression of key proteins including T cell surface glycoprotein CD3 gamma chain (Cd3g) and lymphocyte cytosolic protein 2 (Lcp2). Furthermore, lower expression of cytokines and receptors responsible for lymphocyte proliferation, differentiation, and activation (e.g., Il12r, Il15ra, Il7, Il16, Tgfb1) was observed in the colonic epithelium of the SHR. Finally, Alpi and its product, intestinal alkaline phosphatase, primarily localized in the epithelial cells, were profoundly lower in the SHR. These observations demonstrate that the colonic epithelium undergoes functional changes linked to altered immune, barrier function, and dysbiosis in hypertension.
Collapse
Affiliation(s)
- Tao Yang
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Hongbao Li
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Aline C Oliveira
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Ruby Goel
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Elaine M Richards
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
85
|
Plaeke P, De Man JG, Smet A, Malhotra-Kumar S, Pintelon I, Timmermans JP, Nullens S, Jorens PG, Hubens G, De Winter BY. Effects of intestinal alkaline phosphatase on intestinal barrier function in a cecal ligation and puncture (CLP)-induced mouse model for sepsis. Neurogastroenterol Motil 2020; 32:e13754. [PMID: 31751495 DOI: 10.1111/nmo.13754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Sepsis is a severe pathological condition associated with systemic inflammation, intestinal inflammation, and gastrointestinal barrier dysfunction. Intestinal alkaline phosphatase (IAP) has been demonstrated to detoxify lipopolysaccharide, an important mediator in the pathophysiology of sepsis. We investigated the effect of treatment with IAP on intestinal permeability, intestinal inflammation, and bacterial translocation. METHODS OF-1 mice were divided into 4 groups (n = 12/group), undergoing either a sham or cecal ligation and puncture (CLP) procedure to induce sepsis. Mice received IAP or a vehicle intraperitoneally 5 minutes prior to the onset of the CLP or sham procedure, which was repeated every 12 hours for two consecutive days. After two days, in vivo intestinal permeability, intestinal inflammation, and bacterial translocation were determined. KEY RESULTS CLP-induced sepsis resulted in significantly more weight loss, worse clinical disease scores, bacterial translocation, and elevated inflammatory cytokines. Intestinal permeability was increased up to 5-fold (P < .001). IAP activity was significantly increased in septic animals. Treatment with IAP had no effect on clinical outcomes but reduced the increased permeability of the small intestine by 50% (P = .005). This reduction in permeability was accompanied by a modified gene expression of claudin-1 (P = .025), claudin-14 (P = .035), and interleukin 12 (P = .015). A discriminant analysis showed that treatment with IAP is linked to modified mRNA levels of several tight junction proteins and cytokines. CONCLUSIONS AND INFERENCES Treatment with IAP diminished CLP-induced intestinal barrier disruption, associated with modified expression of several cytokines and claudins. Nevertheless, this effect did not translate into better clinical outcomes in our experimental setup.
Collapse
Affiliation(s)
- Philip Plaeke
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium
- Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium
- Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium
- Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | | | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Sara Nullens
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium
- Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| | - Philippe G Jorens
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium
- Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
- Department of Intensive Care Medicine, Antwerp University Hospital, Edegem (Antwerp), Belgium
| | - Guy Hubens
- Department of Abdominal Surgery, Antwerp University Hospital, Edegem (Antwerp), Belgium
- Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), University of Antwerp, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium
- Infla-Med Research Consortium, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
86
|
Ghosh SS, Wang J, Yannie PJ, Ghosh S. Intestinal Barrier Dysfunction, LPS Translocation, and Disease Development. J Endocr Soc 2020; 4:bvz039. [PMID: 32099951 PMCID: PMC7033038 DOI: 10.1210/jendso/bvz039] [Citation(s) in RCA: 392] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/05/2020] [Indexed: 12/24/2022] Open
Abstract
The intestinal barrier is complex and consists of multiple layers, and it provides a physical and functional barrier to the transport of luminal contents to systemic circulation. While the epithelial cell layer and the outer/inner mucin layer constitute the physical barrier and are often referred to as the intestinal barrier, intestinal alkaline phosphatase (IAP) produced by epithelial cells and antibacterial proteins secreted by Panneth cells represent the functional barrier. While antibacterial proteins play an important role in the host defense against gut microbes, IAP detoxifies bacterial endotoxin lipopolysaccharide (LPS) by catalyzing the dephosphorylation of the active/toxic Lipid A moiety, preventing local inflammation as well as the translocation of active LPS into systemic circulation. The causal relationship between circulating LPS levels and the development of multiple diseases underscores the importance of detailed examination of changes in the “layers” of the intestinal barrier associated with disease development and how this dysfunction can be attenuated by targeted interventions. To develop targeted therapies for improving intestinal barrier function, it is imperative to have a deeper understanding of the intestinal barrier itself, the mechanisms underlying the development of diseases due to barrier dysfunction (eg, high circulating LPS levels), the assessment of intestinal barrier function under diseased conditions, and of how individual layers of the intestinal barrier can be beneficially modulated to potentially attenuate the development of associated diseases. This review summarizes the current knowledge of the composition of the intestinal barrier and its assessment and modulation for the development of potential therapies for barrier dysfunction-associated diseases.
Collapse
Affiliation(s)
| | - Jing Wang
- Department of Internal Medicine, VCU Medical Center, Richmond, Virginia
| | - Paul J Yannie
- Hunter Homes McGuire VA Medical Center, Richmond, Virginia
| | - Shobha Ghosh
- Department of Internal Medicine, VCU Medical Center, Richmond, Virginia.,Hunter Homes McGuire VA Medical Center, Richmond, Virginia
| |
Collapse
|
87
|
Singh SB, Carroll-Portillo A, Coffman C, Ritz NL, Lin HC. Intestinal Alkaline Phosphatase Exerts Anti-Inflammatory Effects Against Lipopolysaccharide by Inducing Autophagy. Sci Rep 2020; 10:3107. [PMID: 32080230 PMCID: PMC7033233 DOI: 10.1038/s41598-020-59474-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/23/2020] [Indexed: 12/21/2022] Open
Abstract
Intestinal alkaline phosphatase (IAP) regulates bicarbonate secretion, detoxifies lipopolysaccharide (LPS), regulates gut microbes, and dephosphorylates proinflammatory nucleotides. IAP also exhibits anti-inflammatory effects in a Toll-like Receptor-4 (TLR-4) dependent manner. However, it is not known whether IAP induces autophagy. We tested the hypothesis that IAP may induce autophagy which may mediate the anti-inflammatory effects of IAP. We found that exogenous IAP induced autophagy in intestinal epithelial cells and in macrophages. TLR4INC34 (C34), a TLR4 signaling inhibitor, suppressed IAP-induced autophagy. IAP also inhibited LPS-induced IL-1β mRNA expression and activation of NF-κB. When autophagy was blocked by 3-methyladenine (3MA) or by Atg5 siRNA, IAP failed to block LPS-mediated effects. IAP also upregulated autophagy-related gene expression in small intestine in mice. We administered either vehicle or IAP (100 U/ml) in drinking water for 14 days in C57BL/6 mice. Mice were sacrificed and ileal tissues collected. Increased expression of Atg5, Atg16, Irgm1, Tlr4, and Lyz genes was observed in the IAP treated group compared to the vehicle treated group. Increase in Atg16 protein expression and fluorescence intensity of LC3 was also observed in IAP-treated tissues compared to the vehicle-treated tissues. Thus, our study lays the framework for investigating how IAP and autophagy may act together to control inflammatory conditions.
Collapse
Affiliation(s)
- Sudha B Singh
- Biomedical Research Institute of New Mexico, VA Health Care System, Albuquerque, New Mexico, USA, 87108
| | - Amanda Carroll-Portillo
- Biomedical Research Institute of New Mexico, VA Health Care System, Albuquerque, New Mexico, USA, 87108
| | - Cristina Coffman
- Biomedical Research Institute of New Mexico, VA Health Care System, Albuquerque, New Mexico, USA, 87108
| | - Nathaniel L Ritz
- Biomedical Research Institute of New Mexico, VA Health Care System, Albuquerque, New Mexico, USA, 87108.,Department of Anatomy & Neuroscience, University College Cork; APC Microbiome institute, University College Cork, Cork, Ireland
| | - Henry C Lin
- Section of Gastroenterology, Medicine Service, New Mexico VA Health Care System, Albuquerque, New Mexico, USA, 87108. .,Division of Gastroenterology and Hepatology, Department of Medicine, the University of New M5052651711exico, Albuquerque, New Mexico, 87131, USA.
| |
Collapse
|
88
|
Peng C, Xing H, Xue Y, Wang J, Li J, Wang E. Ratiometric sensing of alkaline phosphatase based on the catalytical activity from Mn-Fe layered double hydroxide nanosheets. NANOSCALE 2020; 12:2022-2027. [PMID: 31912851 DOI: 10.1039/c9nr08769a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Two-dimensional (2D) Mn-Fe layered double hydroxide (LDH) nanosheets are firstly examined to mediate an O-phenylenediamine (OPD) based fluorescent switch in the presence of ascorbic acid (AA). On one hand, Mn-Fe LDH mimicked the functions of oxidase to catalyze the oxidation of OPD to OPDox, emitting fluorescence at 565 nm. On the other hand, Mn-Fe LDH acts as a superior catalyst for the reaction between AA and OPD to generate 3-(1,2-dihydroxyethyl)furo[3,4-b]quinoxalin-1(3H)-one (N-heterocyclic compound) with an emergence of the maximum emission at 425 nm (ca. 6 fold). The presence of AA not only induces the enhanced emission at 425 nm from the N-heterocyclic compound, but also leads to decreased fluorescence at 565 nm due to the decomposition of Mn-Fe LDH nanosheets. On the basis of the reversed fluorescence response at 425 and 565 nm, ratiometric fluorescence sensing methods (ΔF425 nm/ΔF565 nm) are developed for the determination of AA. With the assistance of alkaline phosphatase (ALP), the activity of ALP can be monitored using the ratiometric platform based on the hydrolyzing ascorbic acid 2-phosphate to yield AA with the detection limit of 0.16 mU mL-1. Different from the traditional ratiometric sensing platform, where two fluorescent probes are often introduced, the present ratiometric system derived from one signal precursor holds great potential in developing a facile platform and broadens the application of 2D nanomaterials in the field of biology.
Collapse
Affiliation(s)
- Chao Peng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | | | | | | | | | | |
Collapse
|
89
|
Identification of a CDH12 potential candidate genetic variant for an autosomal dominant form of transgrediens and progrediens palmoplantar keratoderma in a Tunisian family. J Hum Genet 2020; 65:397-410. [PMID: 31911611 DOI: 10.1038/s10038-019-0711-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 01/10/2023]
Abstract
Molecular diagnosis of rare inherited palmoplantar keratoderma (PPK) is still challenging. We investigated at the clinical and genetic level a consanguineous Tunisian family presenting an autosomal dominant atypical form of transgrediens and progrediens PPK to better characterize this ultrarare disease and to identify its molecular etiology. Whole-exome sequencing (WES), filtering strategies, and bioinformatics analysis have been achieved. Clinical investigation and follow up over 13 years of this Tunisian family with three siblings formerly diagnosed as an autosomal recessive form of Mal de Melela-like conducted us to reconsider its initial phenotype. Indeed, the three patients presented clinical features that overlap both Mal de Meleda and progressive symmetric erythrokeratoderma (PSEK). The mode of inheritance was also reconsidered, since the mother, initially classified as unaffected, exhibited a similar expression of the disease. WES analysis showed the absence of potentially functional rare variants in known PPKs or PSEK-related genes. Results revealed a novel heterozygous nonsynonymous variant in cadherin-12 gene (CDH12, NM_004061, c.1655C > A, p.Thr552Asn) in all affected family members. This variant is absent in dbSNP and in 50 in-house control exomes. In addition, in silico analysis of the mutated 3D domain structure predicted that this variant would result in cadherin-12 protein destabilization and thermal instability. Functional annotation and biological network construction data provide further supporting evidence for the potential role of CDH12 in the maintenance of skin integrity. Taken together, these results suggest that CDH12 gene is a potential candidate gene for an atypical presentation of an autosomal dominant form of transgrediens and progrediens PPK.
Collapse
|
90
|
Wang X, Jiang X, Wei H. Phosphate-responsive 2D-metal–organic-framework-nanozymes for colorimetric detection of alkaline phosphatase. J Mater Chem B 2020; 8:6905-6911. [DOI: 10.1039/c9tb02542a] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Phosphate-responsive peroxidase-mimicking two-dimensional-metal–organic-framework nanozymes were employed to develop alkaline phosphatase assays with tunable dynamic ranges and colorimetric logic gates.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences
- Nanjing National Laboratory of Microstructures
- Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)
- Nanjing University
- Nanjing
| | - Xiaoqian Jiang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences
- Nanjing National Laboratory of Microstructures
- Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)
- Nanjing University
- Nanjing
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences
- Nanjing National Laboratory of Microstructures
- Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Center (ChemBIC)
- Nanjing University
- Nanjing
| |
Collapse
|
91
|
Qi S, Zheng H, Qin H, Zhai H. Development of a facile and sensitive method for detecting alkaline phosphatase activity in serum with fluorescent gold nanoclusters based on the inner filter effect. Analyst 2020; 145:3871-3877. [DOI: 10.1039/d0an00052c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this work, a simple and sensitive method based on the inner filter effect (IFE) of p-nitrophenol (PNP) on the fluorescence of gold nanoclusters (AuNCs) has been developed for detecting alkaline phosphatase (ALP) activity.
Collapse
Affiliation(s)
- Shengda Qi
- College of Chemistry and Chemical Engineering
- Lanzhou University
- Lanzhou 730000
- People's Republic of China
| | - Huanhuan Zheng
- College of Chemistry and Chemical Engineering
- Lanzhou University
- Lanzhou 730000
- People's Republic of China
| | - Hongyan Qin
- Department of Pharmacy
- First Hospital of Lanzhou University
- Lanzhou 730000
- People's Republic of China
| | - Honglin Zhai
- College of Chemistry and Chemical Engineering
- Lanzhou University
- Lanzhou 730000
- People's Republic of China
| |
Collapse
|
92
|
Intestinal Alkaline Phosphatase Deficiency Is Associated with Ischemic Heart Disease. DISEASE MARKERS 2019; 2019:8473565. [PMID: 31915470 PMCID: PMC6930721 DOI: 10.1155/2019/8473565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/20/2019] [Accepted: 11/30/2019] [Indexed: 12/15/2022]
Abstract
Background We have previously shown that the deficiency of the gut enzyme intestinal alkaline phosphatase (IAP) is associated with type 2 diabetes mellitus (T2DM) in humans, and mice deficient in IAP develop the metabolic syndrome, a precipitant of T2DM and ischemic heart disease (IHD). We hypothesized that IAP deficiency might also be associated with IHD in humans. We aimed to determine the correlation between the IAP level and IHD in humans. Methods and Results The IHD patients were recruited from the National Institute of Cardiovascular Diseases (NICVD), Dhaka, Bangladesh, and the control healthy participants were recruited from a suburban community of Dhaka. We determined the IAP level in the stools of 292 IHD patients (187 males, 105 females) and 331 healthy control people (84 males, 247 females). We found that compared to controls, IHD patients have approx. 30% less IAP (mean ± SEM: 63.7 ± 3.5 vs. 44.9 ± 2.1 U/g stool, respectively; p < 0.000001), which indicates that IAP deficiency is associated with IHD, and a high level of IAP is probably protective against IHD in humans. The adjusted generalized linear model (GLM) of regression analysis predicted a strong association of IAP with IHD (p = 0.0035). Multiple logistic regression analysis showed an independent inverse relationship between the IAP level and the IHD status (odds ratio, OR = 0.993 with 95% CI 0.987-0.998; p < 0.01). Conclusions IAP deficiency is associated with IHD, and a high level of IAP might be protective against IHD.
Collapse
|
93
|
Zhang XP, Zhao CX, Shu Y, Wang JH. Gold Nanoclusters/Iron Oxyhydroxide Platform for Ultrasensitive Detection of Butyrylcholinesterase. Anal Chem 2019; 91:15866-15872. [DOI: 10.1021/acs.analchem.9b04304] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xiao-Ping Zhang
- Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, P. R. China
| | - Chen-Xi Zhao
- Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, P. R. China
| | - Yang Shu
- Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, P. R. China
| | - Jian-Hua Wang
- Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, P. R. China
| |
Collapse
|
94
|
Komazin G, Maybin M, Woodard RW, Scior T, Schwudke D, Schombel U, Gisch N, Mamat U, Meredith TC. Substrate structure-activity relationship reveals a limited lipopolysaccharide chemotype range for intestinal alkaline phosphatase. J Biol Chem 2019; 294:19405-19423. [PMID: 31704704 DOI: 10.1074/jbc.ra119.010836] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/31/2019] [Indexed: 12/21/2022] Open
Abstract
Lipopolysaccharide (LPS) from the Gram-negative bacterial outer membrane potently activates the human innate immune system. LPS is recognized by the Toll-like receptor 4/myeloid differentiation factor-2 (TLR4/MD2) complex, leading to the release of pro-inflammatory cytokines. Alkaline phosphatase (AP) is currently being investigated as an anti-inflammatory agent for detoxifying LPS through dephosphorylating lipid A, thus providing a potential treatment for managing both acute (sepsis) and chronic (metabolic endotoxemia) pathologies wherein aberrant TLR4/MD2 activation has been implicated. Endogenous LPS preparations are chemically heterogeneous, and little is known regarding the LPS chemotype substrate range of AP. Here, we investigated the activity of AP on a panel of structurally defined LPS chemotypes isolated from Escherichia coli and demonstrate that calf intestinal AP (cIAP) has only minimal activity against unmodified enteric LPS chemotypes. Pi was only released from a subset of LPS chemotypes harboring spontaneously labile phosphoethanolamine (PEtN) modifications connected through phosphoanhydride bonds. We demonstrate that the spontaneously hydrolyzed O-phosphorylethanolamine is the actual substrate for AP. We found that the 1- and 4'-lipid A phosphate groups critical in TLR4/MD2 signaling become susceptible to hydrolysis only after de-O-acylation of ester linked primary acyl chains on lipid A. Furthermore, PEtN modifications on lipid A specifically enhanced hTLR4 agonist activity of underacylated LPS preparations. Computational binding models are proposed to explain the limitation of AP substrate specificity imposed by the acylation state of lipid A, and the mechanism of PEtN in enhancing hTLR4/MD2 signaling.
Collapse
Affiliation(s)
- Gloria Komazin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Michael Maybin
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Ronald W Woodard
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Thomas Scior
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72000, Mexico
| | - Dominik Schwudke
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Ursula Schombel
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Nicolas Gisch
- Bioanalytical Chemistry, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Uwe Mamat
- Cellular Microbiology, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany
| | - Timothy C Meredith
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
95
|
Regulation of Gut Microbiota and Metabolic Endotoxemia with Dietary Factors. Nutrients 2019; 11:nu11102277. [PMID: 31547555 PMCID: PMC6835897 DOI: 10.3390/nu11102277] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023] Open
Abstract
Metabolic endotoxemia is a condition in which blood lipopolysaccharide (LPS) levels are elevated, regardless of the presence of obvious infection. It has been suggested to lead to chronic inflammation-related diseases such as obesity, type 2 diabetes mellitus, non-alcoholic fatty liver disease (NAFLD), pancreatitis, amyotrophic lateral sclerosis, and Alzheimer’s disease. In addition, it has attracted attention as a target for the prevention and treatment of these chronic diseases. As metabolic endotoxemia was first reported in mice that were fed a high-fat diet, research regarding its relationship with diets has been actively conducted in humans and animals. In this review, we summarize the relationship between fat intake and induction of metabolic endotoxemia, focusing on gut dysbiosis and the influx, kinetics, and metabolism of LPS. We also summarize the recent findings about dietary factors that attenuate metabolic endotoxemia, focusing on the regulation of gut microbiota. We hope that in the future, control of metabolic endotoxemia using dietary factors will help maintain human health.
Collapse
|
96
|
Kaliannan K, Li XY, Wang B, Pan Q, Chen CY, Hao L, Xie S, Kang JX. Multi-omic analysis in transgenic mice implicates omega-6/omega-3 fatty acid imbalance as a risk factor for chronic disease. Commun Biol 2019; 2:276. [PMID: 31372515 PMCID: PMC6659714 DOI: 10.1038/s42003-019-0521-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/28/2019] [Indexed: 12/25/2022] Open
Abstract
An unbalanced increase in dietary omega-6 (n-6) polyunsaturated fatty acids (PUFA) and decrease in omega-3 (n-3) PUFA in the Western diet coincides with the global rise in chronic diseases. Whether n-6 and n-3 PUFA oppositely contribute to the development of chronic disease remains controversial. By using transgenic mice capable of synthesizing PUFA to eliminate confounding factors of diet, we show here that alteration of the tissue n-6/n-3 PUFA ratio leads to correlated changes in the gut microbiome and fecal and serum metabolites. Transgenic mice able to overproduce n-6 PUFA and achieve a high tissue n-6/n-3 PUFA ratio exhibit an increased risk for metabolic diseases and cancer, whereas mice able to convert n-6 to n-3 PUFA, and that have a lower n-6/n-3 ratio, show healthy phenotypes. Our study demonstrates that n-6 PUFA may be harmful in excess and suggests the importance of a low tissue n-6/n-3 ratio in reducing the risk for chronic diseases.
Collapse
Affiliation(s)
- Kanakaraju Kaliannan
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Xiang-Yong Li
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Bin Wang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Qian Pan
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Lei Hao
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Shanfu Xie
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| | - Jing X. Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129 USA
| |
Collapse
|
97
|
Heat stress directly impairs gut integrity and recruits distinct immune cell populations into the bovine intestine. Proc Natl Acad Sci U S A 2019; 116:10333-10338. [PMID: 31064871 PMCID: PMC6535017 DOI: 10.1073/pnas.1820130116] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Consequences of heat stress, particularly for the immune system and the intestinal health of mammals, are a topic of increasing global relevance due to rising temperatures and potential health impairments. Specific climate effects, however, are often difficult to discriminate from indirect consequences, e.g. reduced feed intake. Our study in dairy cattle, which are particularly sensitive to heat, identifies the infiltration of the small intestinal epithelium by a previously unobserved distinct cell population with macrophage-like phenotype in response to moderate heat stress. By using a pair-feeding design, we attributed these effects as direct consequences of heat stress via impaired intestinal barrier function. Therefore, an appropriate gut function is an important component in combating the negative consequences of heat stress. High ambient temperature has multiple potential effects on the organism such as hyperthermia, endotoxemia, and/or systemic inflammation. However, it is often difficult to discriminate between cause and consequence of phenotypic effects, such as the indirect influence of heat stress via reduced food intake. Lactating dairy cows are a particularly sensitive model to examine the effects of heat stress due to their intensive metabolic heat production and small surface:volume ratio. Results from this model show heat stress directly induced a so-far unknown infiltration of yet uncategorized cells into the mucosa and submucosa of the jejunum. Due to a pair-feeding design, we can exclude this effect being a consequence of the concurrent heat-induced reduction in feed intake. Isolation and characterization of the infiltrating cells using laser capture microdissection and RNA sequencing indicated a myeloic origin and macrophage-like phenotype. Furthermore, targeted transcriptome analyses provided evidence of activated immune- and phagocytosis-related pathways with LPS and cytokines as upstream regulators directly associated with heat stress. Finally, we obtained indication that heat stress may directly alter jejunal tight junction proteins suggesting an impaired intestinal barrier. The penetration of toxic and bacterial compounds during heat stress may have triggered a modulated immune repertoire and induced an antioxidative defense mechanism to maintain homeostasis between commensal bacteria and the jejunal immune system. Our bovine model indicates direct effects of heat stress on the jejunum of mammals already at moderately elevated ambient temperature. These results need to be considered when developing concepts to combat the negative consequences of heat stress.
Collapse
|
98
|
Wang F, Li Y, Han Y, Ye Z, Wei L, Luo HB, Xiao L. Single-Particle Enzyme Activity Assay with Spectral-Resolved Dark-Field Optical Microscopy. Anal Chem 2019; 91:6329-6339. [PMID: 30978003 DOI: 10.1021/acs.analchem.9b01300] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In a clinical assay, enzymes are essential biomarkers for human disease diagnosis. In this work, a spectral-resolved single-particle detection (SPD) method is introduced to quantify alkaline phosphatase (ALP) activity in human serum with a supraparticle (SP) based on MnO2-modified gold nanoparticle (denoted as GNP@MnO2 SP) as the probe. In the presence of ALP, 2-phospho-l-ascorbic acid trisodium salt can be hydrolyzed into l-ascorbic acid, which serves as a good reduction agent to trigger the decomposition of the MnO2 shell on the GNP surface. Given that a trace amount of ALP exists, noticeable scattering color change can be detected at the single-particle level due to the sensitive localized surface plasmon resonance (LSPR) effect from GNPs. With spectral-resolved dark-field optical microscopy, a linear dynamic range of 0.06 to 2.48 mU/mL ( R2 = 0.99) and a very low limit of detection of 5.8 μU/mL for the ALP assay are readily achieved, which is more sensitive over the methods based on ensemble sample measurement. As a consequence, this strategy opens a new avenue for the design of an ultrasensitive detection method for disease-correlated biomarker diagnosis in the future.
Collapse
Affiliation(s)
- Fuyan Wang
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research, Ministry of Education, Key Laboratory of Phytochemical R&D of Hunan Province, College of Chemistry and Chemical Engineering , Hunan Normal University , Changsha 410081 , China.,State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin 300071 , China
| | - Yiliang Li
- Department of Rehabilitation Medicine , The Affiliated Baoan Hospital of Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The People's Hospital of Baoan Shenzhen , Shenzhen 510530 , China
| | - Yameng Han
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin 300071 , China
| | - Zhongju Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin 300071 , China
| | - Lin Wei
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research, Ministry of Education, Key Laboratory of Phytochemical R&D of Hunan Province, College of Chemistry and Chemical Engineering , Hunan Normal University , Changsha 410081 , China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , China
| | - Lehui Xiao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin 300071 , China
| |
Collapse
|
99
|
Molecular structure regulation and enzyme cascade signal amplification strategy for upconversion ratiometric luminescent and colorimetric alkaline phosphatase detection. Anal Chim Acta 2019; 1051:160-168. [DOI: 10.1016/j.aca.2018.11.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 12/29/2022]
|
100
|
Zhou W, Davis EA, Dailey MJ. Obesity, independent of diet, drives lasting effects on intestinal epithelial stem cell proliferation in mice. Exp Biol Med (Maywood) 2019; 243:826-835. [PMID: 29932373 DOI: 10.1177/1535370218777762] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The intestinal epithelium plays an essential role in nutrient absorption, hormone release, and barrier function. Maintenance of the epithelium is driven by continuous cell renewal by intestinal epithelial stem cells located in the intestinal crypts. Obesity affects this process and results in changes in the size and function of the tissue. Because both the amount of food intake and the composition of the diet are contributing factors to developing and maintaining obesity, it is necessary to tease apart the separate contributions of obesity versus the type/amount of diet in driving the epithelial changes. C57BL/6J mice were fed a 60% high-fat diet versus a 10% low-fat diet for three months. A pair fed group was included (mice were fed with high-fat diet, but in equal kcal as that eaten by the low-fat diet- fed mice to keep them lean). We investigated the differences in (1) crypt-villus morphology in vivo, (2) the number and function of differentiated epithelial cell types in vivo, and (3) lasting effects on intestinal epithelial stem cell proliferation and growth in vitro. We found that high-fat diet-induced obesity, independent of the high-fat diet, increased crypt depth, villus height, the number of intestinal epithelial stem cells and goblet cells in vivo, and enhanced the size of the enterospheres developed from isolated IESCs in vitro. In addition, there is an interaction of obesity, type of diet, and availability of the diet (pair fed versus ad libitum) on protein and mRNA expression of alkaline phosphatase (an enzyme of enterocytes). These results suggest that high-fat diet-induced obesity, independent of the high-fat diet, induces lasting effects on intestinal epithelial stem cell proliferation, and drives the differentiation into goblet cells, but an interaction of obesity and diet drives alterations in the function of the enterocytes. Impact statement This study investigates whether obesity or the type/amount of diet differentially alters the proliferation, differentiation, growth, and function of the intestinal epithelial tissue. Although diet-induced obesity is known to alter the growth and function of the epithelium in vivo and cause lasting effects in intestinal epithelial stem cells (IESCs) in vitro, we are the first to tease apart the separate contributions of obesity versus the type/amount of diet in these processes. We found that high-fat diet (HFD)-induced obesity, independent of the HFD, drives lasting effects on IESC proliferation and differentiation into goblet cells, which may contribute to the growth of the epithelium. In addition, there is an interaction of obesity, type of diet, and availability of the diet (PF versus ad libitum) on the function of enterocytes. Identification of the factors driving the epithelial changes may provide new therapeutic strategies to control altered tissue growth and function associated with obesity.
Collapse
Affiliation(s)
- Weinan Zhou
- 1 Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Elizabeth A Davis
- 2 Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Megan J Dailey
- 1 Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.,2 Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|